Effects of Human Circulating Hematopoietic Stem & Progenitor Cells on Angiogenesis in the C32 Human Melanoma Xenograft Mouse Model Dillon Etter, Larry.

Slides:



Advertisements
Similar presentations
Immunohistochemistry ESE3 Immunohistochemistry. stained prostate tissue samples for ESE3 troubleshooted ESE3 antibody using the controls - no antibodies.
Advertisements

Cancer-stroma interactions: role of cancer-associated fibroblasts and mast cells in breast carcinogenesis A Malfettone 1, G Simone 2, R Rossi 3, C Salvatore.
Immunohistochemistry
Therapeutic Peptides for Cardiovascular Disease and Cancer Cam Patterson, MD, MBA, FACC, FAHA Ernest and Hazel Craige Distinguished Professor of Cardiovascular.
Na + /H + exchanger regulatory factor 1 (NHERF1) and angiogenesis in familial breast cancer A Mangia*, A Malfettone*, C Salvatore**, B Stea*, G Simone**
Identification of Cancer Stem Cells using Flow Cytometry Analysis تعيين الخلايا الجذعية السرطانية باستخدام تحليل التدفق الخلوي Dr. Ayat Al-Ghafari Biochemistry.
Wijendra Senarathne1, Peggy Gates1, Semir Vranic2, Zoran Gatalica1
Tumor Cell Repopulation between Cycles of Chemotherapy is Inhibited by Regulatory T- Cell Depletion in a Murine Mesothelioma Model  Licun Wu, MD, Zhihong.
The Role of Human Circulating Hematopoietic Stem and Progenitor Cells (CHSPCs) in Promoting Tumor Growth, Angiogenesis and Vascular Repair March 3rd, 2011.
Figure 1. Herbacetin binds to AKT1/2 and suppresses each respective kinase activity. The effect of herbacetin on (A) PI3K/AKT and (B) MAPK signaling pathway.
Corresponding author:
Volume 17, Issue 5, Pages (May 2015)
Infants With Bronchopulmonary Dysplasia Have Fewer Pro-Angiogenic Circulating Progenitor Cells And Decreased Pulmonary Diffusion RS Tepper, C Tiller, J.
Tumor-Induced Sentinel Lymph Node Lymphangiogenesis and Increased Lymph Flow Precede Melanoma Metastasis  Maria I. Harrell, Brian M. Iritani, Alanna Ruddell 
Patient Characteristic
The pCHSPC Fraction Exclusively Stimulates Increased Tube Formation
Figure 1. Herbacetin binds to AKT1/2 and suppresses each respective kinase activity. The effect of herbacetin on (A) PI3K/AKT and (B) MAPK signaling pathway.
Vascular endothelial growth factor-C derived from CD11b+ cells induces therapeutic improvements in a murine model of hind limb ischemia  Go Kuwahara,
1α,25-Dihydroxycholecalciferol and Cyclosporine Suppress Induction and Promote Resolution of Psoriasis in Human Skin Grafts Transplanted on to SCID Mice 
Volume 18, Issue 5, Pages (November 2010)
Direct Comparison of Umbilical Cord Blood versus Bone Marrow–Derived Endothelial Precursor Cells in Mediating Neovascularization in Response to Vascular.
by Alexis S. Bailey, Shuguang Jiang, Michael Afentoulis, Christina I
Overexpression of ornithine decarboxylase enhances endothelial proliferation by suppressing endostatin expression by Takahiro Nemoto, Hisae Hori, Masataka.
Latency-Associated Peptide Prevents Skin Fibrosis in Murine Sclerodermatous Graft- Versus-Host Disease, a Model for Human Scleroderma  Yan Zhang, Laura.
William H. D. Hallett, Weiqing Jing, William R. Drobyski, Bryon D
Myung Jin Son, Kevin Woolard, Do-Hyun Nam, Jeongwu Lee, Howard A. Fine 
Volume 17, Issue 5, Pages (May 2015)
Inhibition of Platelet GPIbα and Promotion of Melanoma Metastasis
The SCF/KIT Pathway Plays a Critical Role in the Control of Normal Human Melanocyte Homeostasis  James M. Grichnik, James A. Burch, James Burchette, Christopher.
Tumorigenic Cells Are Common in Mouse MPNSTs but Their Frequency Depends upon Tumor Genotype and Assay Conditions  Johanna Buchstaller, Paul E. McKeever,
Human endothelial cells express CCR2 and respond to MCP-1: direct role of MCP-1 in angiogenesis and tumor progression by Rosalba Salcedo, Maria Lourdes.
VEGF Gene Delivery to Muscle
Molecular Therapy - Nucleic Acids
Robert E. Merritt, MD, Reiko E. Yamada, BA, Nabil Wasif, MD, Ronald G
Figure SI14. Distribution of compound 1 along with tumor blood vessels
TLR5 signaling in murine bone marrow induces hematopoietic progenitor cell proliferation and aids survival from radiation by Benyue Zhang, Damilola Oyewole-Said,
Volume 5, Issue 5, Pages (November 2015)
by Hairui Su, Chiao-Wang Sun, Szu-Mam Liu, Xin He, Hao Hu, Kevin M
Mesenchymal stem cells expressing osteoprotegerin variants inhibit osteolysis in a murine model of multiple myeloma by Jerome T. Higgs, Joo Hyoung Lee,
SOX2 Is a Marker for Stem-like Tumor Cells in Bladder Cancer
Adhesion receptor expression by hematopoietic cell lines and murine progenitors  Pamela S Becker, Susan K Nilsson, Zhifang Li, Virla M Berrios, Mark S.
Volume 21, Issue 8, Pages (November 2017)
Matthew J. Deeths, Bart T. Endrizzi, Michelle L. Irvin, Lynne P
Volume 16, Issue 3, Pages (September 2009)
Volume 17, Issue 10, Pages (October 2009)
Ravindra Majeti, Christopher Y. Park, Irving L. Weissman 
Interleukin-18 and the Costimulatory Molecule B7-1 Have a Synergistic Anti-Tumor Effect on Murine Melanoma; Implication of Combined Immunotherapy for.
SOX2 Is a Marker for Stem-like Tumor Cells in Bladder Cancer
CD25 expression distinguishes functionally distinct alloreactive CD4+ CD134+ (OX40+) T-cell subsets in acute graft-versus-host disease  Philip R Streeter,
Immunohistochemistry
Volume 15, Issue 3, Pages (March 2009)
Blockade of EDN3/EDNRB signaling impairs GSC self-renewal, cell migration, and tumorigenic capacity. Blockade of EDN3/EDNRB signaling impairs GSC self-renewal,
Volume 15, Issue 1, Pages (January 2009)
Volume 6, Issue 4, Pages (October 2004)
Anti-CD20 CAR exPBNK significantly inhibit growth of Raji cells in xenografted mice. Anti-CD20 CAR exPBNK significantly inhibit growth of Raji cells in.
Common Developmental Pathway for Primitive Erythrocytes and Multipotent Hematopoietic Progenitors in Early Mouse Development  Toshiyuki Yamane, Aya Washino,
In Vivo Gene Therapy with Interleukin-12 Inhibits Primary Vascular Tumor Growth and Induces Apoptosis in a Mouse Model1  Chong Wang, M. Eugenia Quevedo,
Figure 1 A B GCL NBL ED18.5 PND3 PND6 GCL
SLAM Family Markers Resolve Functionally Distinct Subpopulations of Hematopoietic Stem Cells and Multipotent Progenitors  Hideyuki Oguro, Lei Ding, Sean J.
Figure 1 A B GCL NBL ED18.5 PND3 PND6 GCL
Volume 17, Issue 10, Pages (October 2009)
Expression of the Anti-Apoptotic Protein BAG3 in Human Melanomas
Figure 1 A B GCL NBL ED18.5 PND3 PND6 GCL
Figure 1 A B GCL NBL ED18.5 PND3 PND6 GCL
Reversal of diabetic impairments in the blood flow recovery and neovascularization of ischemic areas by ANG-(1-7): A: Representative laser Doppler images.
Induction of MHC-mismatched mixed chimerism depletes plasma and preplasma cells in NOD mice. Induction of MHC-mismatched mixed chimerism depletes plasma.
A, TSG-6 staining in TRAMP tissue and TRAMP cell lines.
Anti-Flk-1 mAb treatment reduces vessel density in tumors.
Alicia G Arroyo, Joy T Yang, Helen Rayburn, Richard O Hynes  Cell 
Molecular Therapy - Nucleic Acids
Presentation transcript:

Effects of Human Circulating Hematopoietic Stem & Progenitor Cells on Angiogenesis in the C32 Human Melanoma Xenograft Mouse Model Dillon Etter, Larry Solomon, George Sandusky, Karen Pollok, and Jamie Case Department of Pathology, Indiana University School of Medicine, Indianapolis Indiana ABSTRACT 7 Days Post-Injection Images 30 Days Post-Injection Images SUMMARY Tumor angiogenesis is a term used to describe the proliferation of blood vessels into a cancerous growth and is an essential process in the metastasis of a tumor. Recent studies suggest that endothelial cells involved in angiogenesis could be derived from bone marrow. Utilizing polychromatic flow cytometry (PFC), we isolated 2 distinct circulating hematopoietic stem and progenitor cell sub-populations. CD31+CD34brightCD45dimAC133+ cells termed pro-angiogenic circulating progenitor cells (PA-CPCs) and CD31+CD34brightCD45dimAC133- cells termed non-angiogenic CPCs (NA-CPCs). Previous findings show that injection of human PA-CPCs into immunodeficient mice with the C32 Human Melanoma Xenograft causes a significant increase in tumor growth, while injection of NA-CPCs does not. This experiment utilized immunohistochemistry techniques to investigate the vascular density at 7 days and 30 days in tumors of mice injected with PA-CPCs, NA-CPCs, and CD34+ cells. Rat anti-mouse CD31 antibody (Dianova) was used to stain mouse endothelial cells in the tumors and Image J software was utilized to quantify the results. At 7 days there was a significant increase in the vascular density, and hence the angiogenic potential of the tumors in mice injected with PA-CPCs compared to those injected with NA-CPCs, CD34+ cells or media. At 30 days, no significant difference in vascular denisty was observed among the four groups of mice. These data suggest that the increased growth of tumors in mice injected with PA-CPCs is due to increased angiogenesis in the early stages of tumor development. Anti-human and anti-mouse CD31, CD34, and CD33 antibodies were used to evaluate vasculature in the tumors. CD31 is an endothelial marker which stains both small and large vessels. CD33 stains myeloid progenitor cells, and CD34 stains adult hematopoietic stem cells. A previous experiment conducted at 28 days post-injection found that there was no human endothelium left in the xenograft after 28 days. Similarly, in this experiment, none of the xenografts harvested at 30 days displayed positive staining for human endothelium. Surprisingly, the xenografts harvested at 7 days were also negative for human cells as evidenced by a lack of anti-human CD31, CD34, and CD33 staining. This data suggests that although human PA- CPCs promote angiogenesis, they are no longer present in the tumor at 7 days. In our quantitative analysis of angiogenesis using a rat anti-mouse CD31 antibody (Dianova), increased staining was observed at 30 days compared to 7 days, suggesting an increase in the amount of murine vasculature between 7 and 30 days. Therefore, although human cells were no longer present at 7 days, the xenografts had not completely vascularized by 7 days. Additionally, out of the 7 day group, those animals that were injected with PA-CPCs exhibited a significant increase in murine vasculature compared to the NA-CPC, CD34+ and media groups. This confirms that PA-CPCs increase tumor angiogenesis. However, results from the 30-day study showed no notable difference in murine vasculature among the four groups of animals, proving that the effect of the human PA-CPCs is no longer visible at 30 days, and that tumors are fully vascularized by this time point. It has recently been reported that xenografts are 50% vascularized by 10 days. The data from this experiment supports that hypothesis. Data from a related experiment showns PA-CPCs also increase the weight and volume of the tumor (Figure 1 & 2). From our observations of xenograft tissues from mice receiving PA-CPCs and NA-CPCs, it was clear that tumors of mice receiving PA-CPCs exhibited less areas of necrosis than those of mice receiving NA-CPCs, CD34+ cells, and media (data not shown). This observation suggests that PA-CPCs could be used as a means of inhibiting necrosis in studies involving the use of xenograft transplants. Media CD34+ Non-Angiogenic CPC Pro-Angiogenic CPC Media CD34+ Non-Angiogenic CPC Pro-Angiogenic CPC BACKGROUND Mouse (+) Control Human (-) Control It has been reported that the ratio of PA-CPCs to NA-CPCs strongly correlates with the severity of the clinical state of patients with peripheral arterial disease (PAD). Patients with PAD exhibit a significant decrease in the ratio of PA-CPCs to NA-CPCs. Conversely, patients with tumor progression exhibit an increase in the ratio of PA-CPCs to NA-CPCs. These data suggest that CD31+CD34brightCD45dimAC133+ PA-CPCs are potential biomarkers for cardiovascular disease and tumor progression. The technique used to isolate these cells is known as PFC, which is used to sort rare cell sub-populations based on the surface markers or antigens they express. A novel PFC protocol has been developed to distinguish and isolate PA-CPCs and NA-CPCs based on AC133 expression. 30 Days Post-Injection Data Figure 1 Figure 2 MATERIALS and METHODS 7 Days Post-Injection Data Tissue specimens: NOD.CB17-Prkdcscid/J (NOD/SCID) mice were subcutaneously injected with 2x106 C32 human melanoma cells and tumor growth was monitored. Once tumors reached ~50mm3, mice were injected with 5x104 PA-CPCs, NA-CPCs, bulk CD34+ cells, or media/vehicle control (PBS). At 7 and 30 days post-injection, mice were euthanized and tumors harvested. Tissue preparation: Tissues were fixed overnight at room temperature in 10% neutral buffered zinc-formalin after which they were transferred through graded concentrations of alcohol to xylene. Tumors were then embedded in paraffin and cut into 6-µm sections. Sections were then mounted onto positively charged slides and baked at 60° C. Immunostaining: Tissues were deparaffinized and hydrated to distilled water. Antigen retrieval was achieved by boiling the slides for 10 minutes in the appropriate buffer (EDTA or citrate, depending on the antibody being used). Endogenous peroxidase activity was blocked using a 3% H2O2 solution. Tissues were incubated with a primary antibody followed by incubation with a biotinylated secondary antibody and avidin-biotin complex (Vector) or with a primary antibody followed by Biocare Mach III polymer kit. Visualization was achieved using DAB chromogen (Sigma). Tissues were counterstained in Mayer’s hematoxylin, dehydrated, and a cover slip was mounted using non-water soluble mounting media. Data Analysis: Images of stained slides at 2.5x magnification were analyzed using Image J software. For small tumors, one field was sufficient to capture a representative portion of the tumor. For large tumors, the average of multiple fields was calculated. Error bars represent the standard error, which was calculated by dividing the standard deviation of a group by the square root of the number of values in that group. CONCLUSIONS The mechanism by which PA-CPCs promote tumor growth is still not completely understood and warrants further investigation. Clarification of the function of these cells may permit their broader application to therapeutic and diagnostic procedures. The results of this study have shown that human PA-CPCs promote tumor angiogenesis and growth in mice harboring C32 Human Melanoma xenografts, and that these human cells are no longer present at 7 days. Furthermore, their pro-angiogenic effects, as indicated by differences in vascular density, are no longer visible by 30 days. Future studies should investigate angiogenesis at time points earlier than 7 days to determine whether the PA-CPCs promote angiogenesis by incorporation into the vasculature of the tumor, or by secretion of pro-angiogenic factors. Group Media CD34+ NA CPC PA CPC Amount of Positive CD31 Staining (Fraction of Low-Power Field) 0.016229098 0.003877163 0.013441358 0.009318775 0.009014428 0.023629583 0.019304854 0.01650121 0.02207757 0.020864198 0.015024599 0.007775422 0.012782735 0.016685258 0.008087355 0.005151721 0.00706326 0.014356354 0.014696736 0.005533563 0.007837648 0.030512875 0.017063065 0.022299383 0.014550365 0.01640872 0.011840578 Average 0.013433418 0.01626405 0.014042904 0.013806928 Standard Deviation 0.00598053 0.008737368 0.0040181 0.006886824 Standard Error 0.002674574 0.00390747 0.001640383 0.001910061 Group Media CD34+ NA CPC PA CPC Amount of Positive CD31 Staining (Fraction of Low-Power Field) 0.00126658 0.006898 0.007175 0.010894 0.00184788 0.003263 0.010159 0.018495 0.008421 0.004968 0.021472 0.003114 0.009379 0.017238 0.010562 0.005337 Mean 0.00155723 0.006452 0.007404 0.017025 Standard Deviation 0.00041104 0.003251 0.002332 0.004456 Standard Error 0.00029065 0.001454 0.001043 0.002228 REFERENCES Sanz, L., et al. (2009). Differential transplantability of human endothelial cells in colorectal cancer and renal cell carcinoma primary xenografts. Laboratory Investigation, 89; 91-97. Estes, M., et al. (2010). Application of polychromatic flow cytometry to identify novel subsets of circulating cells with angiogenic potential.